• Aucun résultat trouvé

Blood–Brain Barrier Dysfunction in a 3D In Vitro Model of Alzheimer's Disease

N/A
N/A
Protected

Academic year: 2021

Partager "Blood–Brain Barrier Dysfunction in a 3D In Vitro Model of Alzheimer's Disease"

Copied!
11
0
0

Texte intégral

(1)

Blood–Brain Barrier Dysfunction in a 3D

In Vitro Model of Alzheimer's Disease

The MIT Faculty has made this article openly available. Please share

how this access benefits you. Your story matters.

Citation

Shin, Yoojin et al. "Blood–Brain Barrier Dysfunction in a 3D In Vitro

Model of Alzheimer's Disease." Advanced Science 6, 20 (August

2019): 1900962. © 2019 The Authors

As Published

http://dx.doi.org/10.1002/advs.201900962

Publisher

Wiley

Version

Final published version

Citable link

https://hdl.handle.net/1721.1/129477

Terms of Use

Creative Commons Attribution 4.0 International license

(2)

FULL PAPER

www.advancedscience.com

Blood–Brain Barrier Dysfunction in a 3D In Vitro Model

of Alzheimer’s Disease

Yoojin Shin, Se Hoon Choi, Eunhee Kim, Enjana Bylykbashi, Jeong Ah Kim, Seok Chung,

Doo Yeon Kim, Roger D. Kamm,* and Rudolph E. Tanzi*

DOI: 10.1002/advs.201900962

1. Introduction

Alzheimer’s disease (AD) is the most common form of age-related dementia, characterized pathologically by deposition of β-amyloid (Aβ) peptides and development of neurofibrillary tangles in the brain.[1] In health, the blood–brain barrier (BBB)

is highly selective, formed by brain endothelial cells (bECs),

Harmful materials in the blood are prevented from entering the healthy brain by a highly selective blood–brain barrier (BBB), and impairment of barrier func-tion has been associated with a variety of neurological diseases. In Alzheimer’s disease (AD), BBB breakdown has been shown to occur even before cognitive decline and brain pathology. To investigate the role of the cerebral vasculature in AD, a physiologically relevant 3D human neural cell culture microfluidic model is developed having a brain endothelial cell monolayer with a BBB-like phenotype. This model is shown to recapitulate several key aspects of BBB dysfunction observed in AD patients: increased BBB permeability, decreased expression of claudin-1, claudin-5, and VE-cadherin, increased expression of matrix-metalloproteinase-2 and reactive oxygen species, and deposition of

β-amyloid (Aβ) peptides at the vascular endothelium. Thus, it provides a

well-controlled platform for investigating BBB function as well as for screening of new drugs that need to pass the BBB to gain access to neural tissues.

3D Alzheimer’s Disease Models

© 2019 The Authors. Published by WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and repro-duction in any medium, provided the original work is properly cited. Dr. Y. Shin, Prof. R. D. Kamm

Department of Mechanical Engineering Massachusetts Institute of Technology

500 Technology Square, MIT Building, Room NE47-321 Cambridge, MA 02139, USA

E-mail: rdkamm@mit.edu

Prof. S. H. Choi, Dr. E. Kim, E. Bylykbashi, Prof. D. Y. Kim, Prof. R. E. Tanzi

Genetics and Aging Research Unit McCance Center for Brain Health

Mass General Institute for Neurodegenerative Disease Department of Neurology

Massachusetts General Hospital Harvard Medical School Charlestown, MA 02129, USA E-mail: tanzi@helix.mgh.harvard.edu

The ORCID identification number(s) for the author(s) of this article can be found under https://doi.org/10.1002/advs.201900962.

Dr. J. A. Kim

Biomedical Omics Group Korea Basic Science Institute Cheongju 28119, Republic of Korea Dr. J. A. Kim

Department of Bio-Analytical Science University of Science and Technology Daejeon 34113, Republic of Korea Prof. S. Chung

KU-KIST Graduate School of Converging Science and Technology Korea University

Seoul 02841, Republic of Korea Prof. S. Chung

School of Mechanical Engineering Korea University

Seoul 02841, Republic of Korea Prof. R. D. Kamm

Department of Biological Engineering Massachusetts Institute of Technology

500 Technology Square, MIT Building, Room NE47-321 Cambridge, MA 02139, USA

Prof. R. D. Kamm

Singapore-MIT Alliance for Research & Technology (SMART) BioSystems and Micromechanics (BioSyM)

Singapore 138602, Singapore

preventing harmful materials in the blood from entering the brain. It also regulates transport and clearance of brain Aβ.[2]

Emerging evidence, however, has shown that the BBB is impaired in AD or patients at risk (i.e., those with mild cognitive impairment), and the dysfunctional BBB has been considered a key factor in the cause and consequences of AD.[3–9]

There-fore, there is tremendous interest in devel-oping cell-based standardized models to closely mimic BBB alterations observed in AD. Such models would facilitate studies into the function of the BBB in AD and the mechanism(s) by which BBB impairment affects AD pathogenesis. They would also provide a valuable platform to screen for drugs that improve BBB function.

Cell culture models to investigate AD-associated BBB impairment have previously been devel-oped using cultures of bECs on transwell inserts, incubated with high concentration of synthesized Aβ peptides for short periods. However, these models lack AD neurons and also fail to reproduce the gradual accumulation of soluble Aβ peptides in the extracellular matrix (ECM) and their transport through the local microenvironment.

(3)

Microfluidic technology has emerged as a powerful tool for studying new multicellular phenomena with precise control of 3D cellular and noncellular microenvironments, integration of multiple functional steps in one experimental platform, and improved imaging capabilities. We previously developed a 3D human neural cell culture model of AD in which human-origin neural progenitor cells (NPCs) expressing amyloid precursor protein (APP) or APP/presenilin 1 (PSEN1) with familial AD (FAD) mutations grow to maturity in a 3D culture system (referred as “3D AD” culture).[10,11] Our 3D AD culture exhibits

key events in AD pathogenesis, including extracellular aggrega-tion of Aβ and accumulaaggrega-tion of hyperphosphorylated tau. How-ever, this model lacks a BBB, and thus, is limited in its ability to study the role of BBB biology and function in AD.

In the present study, we successfully reproduced the 3D AD culture system with an intact vascular wall in a 3D microfluidic platform to study direct effects of the AD pathological microen-vironment on bECs, a major component of the BBB. Our micro-fluidic AD platform mimics the cerebral–vascular interface by reconstituting a tube-shaped bEC barrier that has BBB-like phe-notype and integrates into a 3D AD culture system (referred as our “AD model”). Our AD model successfully mimics several vascular alterations observed in AD patients. These include increased BBB permeability with decreased expression of tight junction proteins, claudin-1 and claudin-5, and adherens junction (AJ) protein, VE-cadherin, increased levels of matrix-metallopro-teinase-2 (MMP-2) and reactive oxygen species (ROS), and aggre-gation of Aβ on the abluminal side of the BBB endothelium. To explore the consequences of the impaired BBB in AD pathogen-esis, we introduced thrombin, a bloodborne toxic enzyme, within the endothelialized channel. Thrombin was able to pass the leaky BBB and exacerbated neuronal loss in AD, which was prevented by decreasing BBB permeability using pharmacological inter-ventions. We demonstrate that our AD model serves as a well-controlled platform to understand physiological and pathological mechanisms of BBB dysfunction in AD and can usefully be employed as a standardized therapeutic drug screening platform.

2. Results

2.1. Development of 3D AD-BBB in the Microfluidic Platform We previously reported a unique strategy for recapitulating AD pathology, namely Aβ-driven neurofibrillary tangles, in a human NPC-derived 3D Matrigel culture system.[10,11] Using

our 3D system, we found that ReNcell VM human NPCs (ReN cells, Millipore) expressing FAD mutations in the APP gene (ReN-GA cells) and APP/PSEN1 genes (ReN-mGAP cells) cause robust extracellular deposition of amyloid plaques and also lead to tauopathy. In the present study, we used the ReN cells derived from the ReN-GA and ReN-mGAP, which collec-tively refer to as ReN-AD cells. As a control, we used wild-type ReN (ReN-WT) cells that do not express FAD mutations.

We developed the AD and WT models by incorporating and coculturing a tube-shaped bEC barrier with 3D-differentiated ReN-AD or ReN-WT cultures in a 3D microfluidic platform. The microfluidic platform consists of five parallel channels fabricated from polydimethylsiloxane (PDMS) elastomer

(Figure 1a). These are grouped into a 3D ReN cell chamber and a BBB chamber, which are positioned on either side of a central “barrier” microchannel (MC) that initially separates the two chambers, but is later filled with hydrogel in order to permit them to interact. The ReN-AD or WT cell chamber consists of two MCs, one that contains neural cell medium and a second that contains the 3D ReN cell culture. The BBB chamber consists of a collagen scaffold MC and a bEC barrier MC (Figure 1a). All MCs are bounded by an array of posts in order to contain the injected solutions within their respective MCs by the action of surface tension.

To compensate for the different maturation periods and culture conditions required by the ReN cells and the bEC monolayer, and also to prevent the potential influence of ReN-AD cells on the bEC barrier during monolayer formation, we developed a simple sequential culture method in which the ReN cell and BBB chambers were separated by the barrier MC, an intervening channel initially filled with air, to allow each cell type to mature independently (Figure 1b).

ReN-AD and ReN-WT cells were individually mixed with Matrigel (1:1 dilution in Dulbecco’s Modified Eagle Medium: Nutrient Mixture F-12 (DMEM/F12) medium supplemented with B27 and heparin) at a density of 107 cells mL−1 and injected

into the 3D ReN cell culture MC. After incubation in 5% CO2 at

37 °C for 30 min, ReN cell differentiation media was added into the ReN cell media MC. ReN cells were differentiated for 5–7 d before initiating bEC barrier formation.

To form the bEC barrier, a 4 mg mL−1 collagen type 1 solution

was loaded into the collagen scaffold MC and allowed to gel. On the next day, hCEMC/D3, a well characterized and established brain endothelial cell line,[12,13] was seeded into the bEC barrier

MC and allowed to attach onto the sidewall of the collagen scaf-fold and bottom and top of the MC. The ReN cell chamber and BBB chamber were maintained separately by keeping the barrier MC filled with air. After the bEC barrier formed a monolayer, the barrier channel was filled with 2 mg mL−1 of pH 7.4

col-lagen type 1 to allow communication between the ReN cells and the bEC monolayer through the ECM via diffusion of soluble factors (Figure 1b; Figure S1, Supporting Information).

Confocal immuno-histochemical analysis using antibodies against microtubule-associated protein 2 (MAP2) and glial fibrillary acidic protein (GFAP) showed that most ReN cells dif-ferentiated into MAP2-positive neurons with some GFAP-pos-itive astrocytes in both the 3D ReN-AD and ReN-WT cultures (Figure 1c,d; Figure S2, Supporting Information), confirming successful neuronal differentiation of ReN cells in the 3D ReN culture MC. Enzyme-linked immunosorbent assay (ELISA) analysis confirmed the secretion of Aβ40 and Aβ42, which were detected in the medium of the ReN cell MC, by ReN-AD cul-tures in our AD model (Figure 1e).

Expression and localization of AJs and tight junctions (TJs) of the bEC monolayer were verified with immunofluorescent staining for VE-cadherin (VE-cad, an AJ protein), and ZO-1 and claudin-5 (both TJ proteins). Our staining analysis confirmed that VE-cad, ZO-1, and claudin-5 were expressed and localized along the cell junction of the bEC monolayer (Figure 1f). We also con-firmed the expression of Aβ transporters, receptor for advanced glycation end products (RAGE), P-glycoprotein, and lipoprotein receptor-related protein-1 (LRP1) across the bECs (Figure 1g).

(4)

www.advancedsciencenews.com www.advancedscience.com

To further explore the mechanism of Aβ accumulation in the AD model,[14] a simulation was performed using

COMSOL Multiphysics. Secretion of Aβ and gradual accu-mulation in the ECM in the AD model was simulated as a constant and distributed source of soluble Aβ secreted from ReN-AD cells at a rate of 1.4 × 10−12 mol m−3 s−1 (5 × 10−12m

per hour) (gray box in Figure 2a). Based on the size of soluble Aβ (≈4 kDa), the diffusion coefficient of the bEC barrier was set to 2 × 10−15 m2 s−1, based on the detected

amount of soluble Aβ from media in ReN cell media and bEC monolayer MCs (Figure S3, Supporting Information). Daily medium replacement in the bEC barrier MC was also included. All simulation details are presented in the Experi-mental Section.

2.2. BBB Permeability Was Increased in AD Model

We first tested whether our AD models mimic the BBB break-down observed in AD by directly measuring BBB permeability. Permeability was determined by adding culture medium with 10 × 10−6 m fluorescent (FITC)-labeled or Texas Red-labeled

dextran (Mw: 3 and 40 kDa) in culture media to the bEC

bar-rier MC on days 3 and 6 of coculture, and then imaging the fluorescence every 5 min for more than 30 min. The perme-ability was calculated by measuring the mean fluorescence intensity in a control volume (CV) defined in the collagen gel immediately after introducing the dextran to the MC and again 10 min later, then applying a permeability equation (Figure 2b; Figure S4, Supporting Information). Our BBB permeability Figure 1. Schematic depiction of a WT/AD model and verification of function of 3D ReN culture and BBB integrity. a) The WT/AD model is composed

of ReN-WT/AD cell and BBB chambers with an intervening barrier microchannel (MC). The ReN cell chamber is composed of a ReN cell medium MC and 3D ReN culture MC, and the BBB chamber is composed of a collagen scaffold MC and bEC barrier MC. Numbers (mm) indicate the dimen-sion of our microfluidic model. b) Sequential experimental protocol for WT/AD model development. The mixture of ReN-WT or AD cells in Matrigel (MAT) was injected in 3D ReN culture MC, and ReN differentiation medium (DIFF medium) was added in ReN cell media MC. After 3 d, collagen type 1 (4.0 mg mL−1) was injected in the collagen scaffold MC, followed by MAT coating (1:50 in serum free media). Next day, bECs (hCMEC/D3) were

seeded in the bEC barrier MC, and allowed to form a monolayer on the collagen scaffold and bottom and top of the MC. After monolayer formation, ReN cell and BBB chambers were connected by injecting collagen (2.0 mg mL−1) into the barrier MC. c) 3D-differentiated GFP labeled ReN cells in 3D

AD culture MC, and d) the representative image of 2-week differentiated ReN-AD cells that are stained with MAP2 (red) and an overlay image. Scale bars: 80 µm in (c) and 40 µm in (d). e) Levels of soluble Aβ40 and Aβ42 detected in media from EC only, ReN-WT, or ReN-AD cells in the WT/AD models. f) Expression of junction protein (VE-Cad) and tight-junction proteins (ZO-1 and Claudin-5), and g) Aβ transporters (RAGE, PgP, and LRP-1) in the bEC barrier of the AD model. The expression of tight junction proteins and Aβ transporters was confirmed by immunofluorescence staining. Scale bars: 40 µm. Data are mean ± S.E.M. Statistical analysis was by Student’s t test.

(5)

assay indicated that the permeability of the bEC barrier was significantly increased in the AD model (1.4 × 10−5 cm s−1 for

3 kDa, and 6.45 × 10−6 cm s−1 for 40 kDa), as compared to the

WT model (7.46 × 10−6 cm s−1 for 3 kDa, and 1.96 × 10−6 cm s−1

for 40 kDa) (Figure 2c; Figure S5, Supporting Information).

2.3. Increased BBB Permeability Was Accompanied by Reduced Expression of Claudin-1, Claudin-5, and VE-cadherin in the bEC of AD Models

We next investigated whether increased BBB permeability was accompanied by disruption of the AJs and TJs of the bEC bar-rier of the AD models. We screened for AJ and/or TJ gene expression in the bECs of AD models using qRT-PCR analysis. Since it is difficult to collect sufficient numbers of bECs from the 3D AD/WT models for quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis, ReN-AD or ReN-WT cells were cultured and differentiated on a 24-well plate for 5–7 d, and then combined with a bEC monolayer formed on collagen and matrigel-coated inserts. At coculture day 3, the bECs were collected, and gene expression levels of TJs [CLND1 (claudin-1), CLND5 (claudin-5), OCLN (occludin), and TJP1 (ZO-1)], and the AJ [CDH5 (VE-cadherin)] were analyzed by qRT-PCR. Among them, the levels of CLDN1 (claudin-1), CLDN5 (claudin-5), CDH5 (VE-cad) expressions in bECs were significantly decreased when cocultured with ReN-AD cells compared to those cocultured with ReN-WT cells (Figure 2d).

The levels of OCLN (occludin) and TJP1 (ZO-1) were not significantly different between WT and AD models. To confirm the reduced expression of tight junction proteins in our AD models, the bEC barrier of AD and WT models was fixed on day 3 of coculture and immunostained with antibodies against claudin-5 and VE-cad. Similar to our qRT-PCR data, expression levels of claudin-5 and VE-cad, as viewed from the luminal side of the bEC monolayer formed on the scaffolds in the bEC bar-rier MC, were reduced at cell–cell junctions in the AD model, compared to the WT model (Figure 2e; Figure S6, Supporting Information). These results suggest that the increase of BBB permeability in our 3D AD model is, at least in part, induced by reduced expression of tight junction and adherens proteins, including claudin-1 and claudin-5, and VE-cad.

2.4. Levels of ROS, MMP-2, and Interferon γ (IFNγ) Were Increased in the AD Models

Mechanisms by which BBB is impaired in AD should be diverse and complex, though they are currently not clear. Vascular oxi-dative stress has been suggested to be one of the pathological features that appears in the brains of AD patients.[15–17] Aβ has

been shown to produce severe damage to the endothelial lining through deleterious effects on endothelial nitric oxide (NO)[18,19]

and accumulated oxidative stress interferes with increases of endothelial permeability.[20] Finally, the effect of Aβ has been

attributed to enhanced NO catabolism due to an increased Figure 2. Permeability increase in the AD model. a) The concentration profiles of soluble Aβ secreted from ReN-AD cells in the AD model. Gray shading indicates the level of accumulated Aβ within ECM and the level of Aβ near the bEC barrier is indicated in the yellow bar. b) Permeability values of the bEC barrier in the WT/AD model measured by introducing bEC medium supplemented with 10 × 10−6m dextran (40 kDa (FITC)) into the bEC barrier

MC and monitoring fluorescence every 5 min after flow stabilization. The images were recorded initially (top) and at 600 s (bottom) after adding the dextran. Scale bars: 100 µm. c) Permeability values of the bEC barrier in the WT model and AD model at coculture day 3 (n = 13 for (with WT) and

n = 25 for (with AD) from 3 independent experiments, ****p < 0.0001). d) Relative gene expression levels of tight junction proteins of claudin-1 and claudin-5 (CLDN1 & 5), occludin (OCLN), and ZO-1 (TJU1) and adherens junction protein VE-cadherin (CDH5) expressed in the bEC barrier in the WT model and AD model (n = 6, *p < 0.05, ***p < 0.001, biological replicates = 3). e) Comparison of tight junction protein expression in the bEC barrier cultured with WT and AD cells in the WT/AD model. The junction protein expression level of cluadin-5 was visualized by immunofluorescence staining with an antibody against claudin-5 with DAPI staining for cell nuclei. Graphs show that intensity profiles of distribution of claudin-5 peaking at the cell junctions (arrows). Each colored line indicates the intensity profile of claudin-5 expressed by randomly selected cells. Scale bars: 30 µm. Data are mean ± S.E.M. Statistical analysis was by Student’s t test.

(6)

www.advancedsciencenews.com www.advancedscience.com production of ROS.[21,22] Therefore, we measured intracellular

ROS levels in the bECs using CellROX orange reagent at cocul-ture day 3. As shown in Figure 3a, we observed that ROS is significantly higher in the bEC barrier of 3D AD models com-pared to 3D WT models. We also observed an increase in the number of caspase 3/7-positive apoptotic bECs in the 3D AD model compared to the 3D WT model (Figure 3b).

MMPs are a family of enzymes able to degrade components of the extracellular matrix, which are important for normal BBB function. MMP-2 has been demonstrated to be a key element in the induction of BBB permeability alterations and disrup-tion of TJs.[23–25] It is secreted by ECs in cultures upon

stimu-lation with Aβ peptides.[26] AD patients showed higher plasma

MMP-2 levels, although contradicting results have also been reported.[27,28] We measured the levels of secreted MMP-2,

MMP-3, and MMP-9 from day 2 to day 6 of coculture in the bEC media of AD and WT models. Among them, the levels of MMP-2 were significantly higher in the 3D AD model com-pared to the 3D WT model (Figure 3c). We did not observe any change in the level of MMP-3 between the two models, and MMP-9 was not detectable in either model.

In addition, inflammatory cytokines have been known to contribute to the increase of BBB permeability.[29–31] To

deter-mine if the level of inflammatory cytokines increases in our AD model, we measured the levels of IFNγ, interleukin-1β (IL-1β), IL-6, IL-8, and transforming growth factor α (TNFα), all of which are associated with the increase of BBB permeability, from day 2 to day 6 in the bEC media of our two models. We found that the level of IFNγ detected over 5 d was significantly higher in the media of 3D AD models compared to that of 3D

WT models (Figure 3d). Since we did not detect IFNγ in the EC culture medium (data not shown), the detected level of IFNγ is most likely secreted from ReN-AD cells and had crossed into the bEC barrier. Interestingly, IFNγ has been shown to trigger ROS production[32–34] and also contribute to the increase of

BBB permeability.[35,36]

2.5. Reducing Aβ Generation Decreased BBB Permeability To explore whether reducing Aβ generation ameliorates BBB impairment, we treated ReN-AD cells with BACE1 inhibitor (LY2886721), which prevents the production of Aβ by blocking the β-secretase enzyme. We added 1 × 10−6m of LY2886721 in

the ReN cell differentiation media MC. LY2886721 at this dose dramatically reduced the levels of Aβ40 and Aβ42 secreted from ReN-AD cells compared to vehicle (0.01% dimethyl sulfoxide (DMSO)) (Figure 3e). We performed a permeability measure-ment by introducing 3 kDa and 40 kDa FITC dextrans into the bEC barrier MC on day 3 of coculture. The treatment of LY2886721 significantly decreased BBB permeability (Figure 3f) and increased the level of CLDN5 expression in bECs (Figure 3g). These results suggest that Aβ itself and/or Aβ-driven changes in other molecules play a role in causing BBB impairment. 2.6. Deposition of Aβ on the bEC Barrier

Aβ deposition is observed in the cerebrovasculature and is usually described as cerebral amyloid angiopathy (CAA). Aβ

Figure 3. The increased levels of ROS, MMP-2, and IFNγ in the AD model. a) Relative mean fluorescence intensity of level of intercellular reactive oxygen species (ROS) in the bEC barrier. The level of ROS was confirmed by CellROX Orange. Representative images show that the bEC barrier cocultured with AD cells expressed higher levels of intercellular ROS (indicated with red staining) than those cocultured with WT cells (n = 10–13. Biological replicates = 3, **p < 0.01). Scale bars: 70 µm. b) Apoptosis of bECs were detected by CellEvent Caspase-3/7 Green Detection Reagent and quantified by counting caspase 3/7 (green) positive cells. Representative images show that the bEC barrier cocultured with AD cells has more caspase 3/7 (green) positive cells. Scale bars: 40 µm (n = 8 for WT, n = 16 for AD, biological replicates = 3, *p < 0.05). c) The level of soluble MMPs including MMP-2, MMP-3, and MMP-9 in bEC media in the WT/AD models (n = 11–14. Biological repeat = 2, *p < 0.05, n.s., not significant, #, not detected). d) The level of inflammatory cytokines in the WT/AD model (n = 8–9, **p < 0.01). e) The levels of Aβ 40 and Aβ 40 secreted by WT cells, AD cells, and AD cells treated with BACE1 inhibitor (LY-2 886 721) (n = 8, ***p < 0.001). f) Permeability of the bEC barrier in the AD model treated with DMSO and BACE 1 inhibitor (LY2886721) (n = 16–19, biological replicates = 5, ***p < 0.001, ****p < 0.0001). g) Relative gene expression level of tight junction proteins of claudin-5 (CLDN5) expressed in the bEC barrier in the AD model with or without LY2886721 treatment (n = 3–5, *p = 0.0135). Data are mean ± S.E.M. Statistical analysis was by Student’s t test.

(7)

deposition in the vasculature leads to proinflammatory and cyto-toxic events that contribute to the greater BBB permeability in the AD brain. To check whether Aβ species secreted from ReN AD cells are directly deposited onto the bEC barrier in our AD model, we performed immunofluorescent staining with Aβ40 and Aβ42 antibodies at coculture day 6. Aβ deposition was evi-dent in the 3D AD model, but not in 3D WT model (Figure 4a). The total amount of Aβ deposition on the surface of the bEC barrier of our 3D AD model was not extensive, however, pos-sibly due to the short period (7 d) of bEC and ReN cell coculture. To explore which Aβ species, Aβ40 or Aβ42, dominantly depos-ited on the BBB wall, we introduced Fluor 555-labeled Aβ40 and FAM-labeled Aβ42 together in the ReN AD cell media MC and found that both Aβ40 and Aβ42 were codeposited (Figure 4b). These results demonstrate that our AD models successfully mimic the Aβ deposition patterns of CAA around the blood ves-sels of the brain and might therefore also be useful for studying other functional consequences associated with CAA.

In order to explore whether Aβ can pass the BBB once its permeability is increased, we measured the levels of Aβ40 and Aβ42 in the bEC barrier MC. Although the levels of both were higher in the 3D ReN cell MC of the AD model compared to the WT model (Figure 1e), they were below the detectable range in

the bEC barrier MC (Figure S3 of the Supporting Information, left graphs). Thus, it appears that Aβ species deposit and aggre-gate on the surface of the BBB in our models rather than pass through the disrupted BBB.

2.7. The Effects of BBB Disruption on Neuronal Damage In AD patients, it is possible that circulating neurotoxins enter the brain through the disrupted BBB and exacerbate AD progression. Therefore, enhancing BBB integrity by pharmaceutical drugs might yield beneficial therapeutic effects by blocking the neuro-toxins from entering the brain. We tested these hypotheses using our AD models. We first demonstrated that the leakage of neuro-toxins through the disrupted BBB can damage neural cells. For this, we introduced thrombin, a serine protease which is directly neurotoxic, that causes proinflammatory effects and whose level is elevated in the brain and cerebral microvasculature in AD,[37]

into the bEC barrier MC on coculture day 3 (Figure 5a). After 3 h of treatment, cell death as determined by ethidium homodimer-1, increased in the AD models (Figure 5b; Figure S7, Supporting Information). This suggests that thrombin passed through the disrupted BBB to the AD cultures and exacerbated cell death.

Next, we tested whether the disrupted BBB in our system can be restored by pharmacological compounds that enhance BBB integrity. It has been shown that etodolac and beclometha-sone have the potential to increase BBB integrity against amy-loid toxicity.[38] We treated the bEC barrier of AD models with

either etodolac or beclomethasone at EC50 concentrations of 2.32 and 0.76 × 10−6m, respectively, on coculture day 2. After

24 h, we performed a permeability analysis by introducing 3 and 40 kDa dextran into the bEC barrier MC. Etodolac signifi-cantly decreased the permeability of the bEC in the AD model compared to vehicle (0.01% DMSO), while beclomethasone did not show such effects (Figure 5c). Etodolac also significantly increased the expression of claudin-5 localized between cell–cell junctions of the bEC barrier (Figure 5d). Therefore, etodolac, but not beclomethasone, effectively restored the integrity of the impaired bEC barrier in the AD model.

In order to explore whether increasing BBB integrity phar-macologically has beneficial effects on thrombin-induced cell death in AD, we treated our AD models with etodolac for 24 h on coculture day 2 and introduced thrombin into the bEC bar-rier MC on day 3. We observed that the level of dead cells in the 3D AD model treated with etodolac was significantly lower compared to treatment with vehicle (0.01% DMSO) (Figure 5e), leading to the suggestion that drugs that increase BBB integrity might have therapeutic potential for AD and that our 3D AD models can be used to screen those drugs.

3. Discussion

Numerous studies have suggested that BBB breakdown initi-ates and/or contributes to a vicious cycle of AD progression, resulting in neuronal loss and dysfunction.[3–5,7–9] Because of

the complexity of the in vivo BBB, simplified in vitro models have been developed that mimic barrier impairment observed in AD and offer considerable potential to provide insights into Figure 4. Aβ deposition in the AD model. a) Deposition of Aβ (green)

on the BBB monolayer determined by immunofluorescence staining with Aβ 42/40 antibody. White arrows indicate deposited Aβ on the bEC bar-rier. Scale bars: 30 µm. b) Deposition of flour 555-labeled Aβ 40 and FAM-labeled Aβ 42 which were introduced into ReN cell media MC, on the bEC barrier. White arrows indicate deposited Aβ on the bEC barrier. Scale bars: 50 µm.

(8)

www.advancedsciencenews.com www.advancedscience.com

AD pathology and to aid in the development of new drugs for the disease. AD-associated BBB impairment in a cell culture system has been studied by culturing bECs on transwell inserts and by treating the bECs with high concentrations of synthe-sized Aβ peptides for short periods.[39–41] However, these classic

models lack many features of the human brain in AD and have therefore been of limited value. In the present study, we suc-cessfully developed a more physiologically relevant 3D human neural cell culture model of AD with a bEC barrier in a micro-fluidic platform and verified that it successfully mimics certain aspects of bEC barrier dysfunction, notably the increase in per-meability observed in human AD patients.

Our AD model has the following capabilities, which were otherwise difficult to perform with the traditional in vitro

BBB transwell models: 1) it provides a 3D microenviron-ment for cells to quantitatively evaluate bEC barrier perme-ability; 2) it allows for integration of multiple functional steps in a single experimental platform; 3) it enables enhanced imaging capabilities and economy in reagent and tissue sample size; 4) the developed sequential, compartmental-ized culture method allows for the different culture condi-tions and maturation periods of ReN cells and the bECs; and 5) it realizes a gradual accumulation of Aβ appearing in brains of AD patients by allowing AD cells to continually release Aβ, which gradually accumulates within and diffuse into the ECM. These capabilities enable more physiologically relevant in vitro modeling in our AD model compared with other previous in vitro models.

Figure 5. Demonstration of influx of neurotoxins through the impaired BBB and its effects on neural cell damage, and a possible application of drug

evaluation for a BBB enhancer. a) Neural cell damage by thrombin inflow through the impaired bEC barrier. Cell death was assayed using ethidium homodimer-1 after introducing thrombin into the bEC barrier MC at coculture day 3. Dead cells were shown in red. Scale bars: 200 µm. b) The number of dead cells (red) in the ROI (in 0.73 mm2) (n ≥ 9 for WT/AD with thrombin; n ≥ 4 for AD without thrombin, **p < 0.01, ****p < 0.0001). c)

Perme-ability of the bEC barrier cocultured with AD cells with or without drug treatment (n ≥ 26 for 0.01 % DMSO and etodoalc, n > 5 for beclomethasone, ***p < 0.001, n.s., not significant). d) Comparison of tight junction protein expression in the bEC barrier of the AD model after drug treatment for 24 h. The junction protein expression of claudin-5 was visualized by immunofluorescence staining with a claudin-5 antibody and DAPI for cell nuclei. Graphs show the intensity profile of claudin-5 at the cell junctions. Arrows indicate claudin-5 expressions at the junctions of cells. Each colored line indicates the intensity profile of claudin-5 expressed by randomly selected cells. Scale bars: 20 µm. e) Comparison of the amount of dead (damaged) WT/AD neural cells in the 3D ReN culture MC. Cell death was analyzed by counting dead cells (red) in ROI (in 0.73 mm2) (n ≥ 16, ****p < 0.0001).

(9)

The paracellular permeability of the bEC barrier was increased in our AD models and it was accompanied by decreased expression of tight junction proteins, claudin-1 and claudin-5, and an adherens junction protein, VE-cadherin. The mechanism(s) of BBB impairment in AD are likely diverse and complex. Aβ might directly increase permeability of the bEC barrier. We found that the levels of oxidative stress, MMP-2, and IFNγ were increased in our AD model compared to the WT model, all of which have shown to impair BBB function in AD. Interestingly, Aβ did not pass the impaired BBB, but rather deposited on the wall of bECs, mimicking CAA pathology. Future studies will require investigation of whether expression of Aβ transporters is altered on the bECs of our AD model. Reducing Aβ production by the treatment of BACE1 inhibitor, LY-2 886 721, significantly reduced the permeability of bEC barrier in our model, while it increased the expression level of claudin-5. These results suggest that Aβ itself and/or Aβ-driven molecular changes effect BBB dysfunction in AD and that reducing parenchymal Aβ pharmacologically could also reduce BBB leakage in AD.

Using our AD model, we were also able to suggest that increasing BBB integrity pharmacologically might prove to be a useful therapeutic approach for AD treatment by preventing blood-originated neurotoxins from entering the brain. Accumu-lation of neurotoxins, such as fibrinogen and thrombin, that enter though impaired BBB from the circulation was observed in AD patients.[42,43] We first demonstrated that the leakage of

neurotoxins through a disrupted BBB exacerbates neuronal death by allowing human thrombin (≈38 kDa) transport across the bEC barrier. These results support the hypothesis that accumulation of neurotoxins in the brains can exacerbate AD pathogenesis.[37,44] We then showed that enhancing BBB

integ-rity by etodolac significantly decreased the death of ReN-AD cells induced by thrombin entry. The BBB enhancing effect of beclomethasone was not observed in the AD model, while it sig-nificantly enhanced BBB integrity in the transwell model with synthesized Aβ mixture.[38] This difference in drug effect might

be due to the fact that bECs dysfunction in our AD model is affected by not only Aβ peptides but also by other pathological environmental stimuli, such as cytokines from AD cells. If so, it suggests that our model might more realistically replicate in vivo behavior and hence serve as a useful drug screening tool.

We recognize that we do not reconstitute the BBB vasculature entirely. Our model does not contain astrocytes and pericytes both of which play roles in BBB maintenance and function. Rather, in the present study, we have chosen to first study a simpler dis-ease model in order to focus on interactions between the AD cells and bECs, both key components of the BBB. In addition, it has recently been reported that the cerebral vasculature of AD patients has 50% less coverage of pericytes than healthy brains,[45]

and also astrocyte degradation is observed in AD,[46] which

sug-gests that bECs might be more likely to be directly exposed to environmental toxic stimuli of AD. Therefore, our models will be important in the study of changes in vascular endothelium itself or in its role in AD progression, and how BBB properties are dic-tated by intrinsic features of bECs in AD pathogenesis.

Recently, attempts have been made to develop more human-relevant BBB cultures in vitro by employing induced pluripotent stem cells (iPSCs).[47–50] However, iPSCs are donor-specific, and

thus highly variable in their behavior, so it would be difficult to develop a standardized BBB model based on iPSCs for drug discovery and development. We chose, therefore, to use well-characterized and established cell lines, such as hCEMC/D3, in order to develop stable and standardized assays for those pur-poses with better quality assurance/qualify control (QA/QC).

We believe that our AD model will be a useful standard-ized tool in the study of BBB biology in AD and also provide a platform for moderate-throughput drug screening of drugs to inhibit BBB dysfunction or enhance BBB integrity as an adjunct to other AD therapies.

4. Experimental Section

Fabrication of Microfluidic Platform: The micropatterned master

with features 150 µm in height was fabricated by using standard photolithography. SU-8 100 was coated and patterned on a 4 in. silicon wafer by exposing UV light to the top of a transparent photomask on which the desired patterns are printed. A mixture of PDMS (Sylgard 184, Dow-Corning, Midland, MI, USA) and its curing agent were mixed to a 10:1 weight ratio, poured onto the patterned master to a 5 mm thickness, degassed to remove air bubbles, and cured in an oven at 80 °C for 1.5 h. The cured PDMS was removed from the wafer and holes were punched through the reservoir patterns to make inlets and outlets for media and hydrogel filling. After sterilization, the devices and glass coverslips were assembled by using oxygen plasma treatment and filled with 60 µL of 1 mg mL−1 poly (D-lysine) solution (PDL, M.W. 70 000–150 000, Sigma-Aldrich, St. Louis, MO, USA) to strengthen the attachment of hydrogel to the surface. After incubation for 2 h at 37 °C, the PDL solution was removed, and then the devices were rinsed with water and dried at 80 °C overnight.

Cell Culture: Human cerebral microvascular endothelial cells

(hCMEC/D3, Cedarlane, Ontario, Canada), a cell line derived from human temporal lobe microvessels, were cultured to 90% confluence on a 150 µg mL−1 collagen type 1 coated flask in EndoGRO-MV Complete

Culture Media Kit (Millipore, Temecula, CA, USA) supplemented with 1 ng mL−1 bFGF (R&D System, Inc., Minneapolis, MN, USA) and 1%

penicillin-streptomycin. ReN-WT and ReN-AD cells were cultured on a Matrigel coated flask in proliferation media; DMEM/F12 (Gibco/Life Technologies, Gaithersburg, MD, USA) supplemented with B27 (Life Technologies, Carlsbad, CA, USA), heparin (2 mg mL−1 stock, STEMCELL

Technologies, Carlsbad, CA, USA), 20 ng mL−1 epidermal growth factor

(EGF), and 20 ng mL−1 fibroblast growth factor 2 (FGF-2).

3D Cell Culture in the Microfluidic Platform: ReN-WT and ReN-AD

cells in a 25T confluent flask were washed with D-PBS and treated with Accutase (Life Technologies, Carlsbad, CA, USA) in a 37 °C CO2 incubator for 3–5 min for dissociation of cells. The prewarmed

differentiation medium was added to the flask, and the cell suspensions were transferred into 15 mL tubes and centrifuged at 2000 g for 3 min. The cells were resuspended at a density of 2 × 107 cells mL−1 in the

differentiation medium and placed on ice. ReN cell suspensions were mixed with cold Matrigel at the volume ratio of 1:1 (final density: 1 × 107 cells mL−1), introduced into the 3D AD culture MC, and placed

in a 37 °C CO2 incubator for 30 min. After 30 min, the prewarmed

differentiation medium was injected into the ReN cell media MC. After 5–7 d, 4 mg mL−1 collagen type 1 solution, which was prepared by diluting high concentration collagen type 1 (rat tail, cat. No. 354 249, Corning, NY, USA) in 10× PBS and sterilized deionized water and titrated to pH 7 with 0.5 N NaOH, was injected into the collagen scaffold MC and incubated at 37 °C and 5% CO2 for 30 min. After 1 d, bEC barrier

MC was coated with growth factor reduced Matrigel (1:50 diluted in serum free media, Corning, NY, USA) for 40 min and hCMEC/D3 cells were seeded at a density of 2 × 106 cells mL−1 on the sidewall of the

collagen scaffold and top and bottom surfaces of the bEC barrier MC. hCMEC/D3 cells were allowed to form a tight monolayer along the MC for 2–3 d in hCMEC/D3 growth medium. After bEC barrier formation,

(10)

www.advancedsciencenews.com www.advancedscience.com

ReN cell chamber and BBB chamber were connected and allowed to communicate by filling 2 mg mL−1 of pH 7 collagen gel (rat tail, cat.

No. 354 249, Corning, NY, USA) into the barrier MC. After connection, hCMEC/D3 cells were cultured in EndoGRO-MV Complete Culture Media Kit (Millipore, Temecula, CA, USA) supplemented with 0.5% fetal bovine serum (FBS), 1 ng mL−1 bFGF (R&D System, Inc., Minneapolis,

MN, USA), and 1% penicillin–streptomycin.

Permeability Measurement: The permeability was determined by

adding hCMEC/D3 culture medium with 10 × 10−6 m FITC-labeled

dextran (Mw: 40 kDa) into the bEC barrier MC 3 and 6 d after connecting

the ReN cell chamber and the BBB chamber. Fluorescent images were recorded every 5 min for 2 h with a confocal microscope. The permeability was estimated by measuring the mean fluorescence intensity in a CV defined in the collagen gel at time 0 and time 600 s and then applying the equation

P VAg 1/ I ddtI v

(

m

)

g

= ∆ (1)

where Vg is gel volume in the CV, Av is vessel surface area in CV, Ig

is fluorescence intensity in gel, Im is fluorescence intensity at the

monolayer (Im = Ilumen − Igel) at time 0, and ddtIg is the rate of increase in

intensity as the dextran diffuses out of the bEC barrier MC into the gel (Figure S4, Supporting Information).

Reconstitution and Treatment of Aβ40 and Aβ42: Vacuum-dried synthetic monomeric Aβ40 and Aβ42, which were obtained from Dr. Robert Moir (Massachusetts General Hospital), were reconstituted by adding 200 µL of 20 × 10−3 m NaOH, mixing gently multiple

times then allowing 30 s for settling. After several mixing steps, the reconstituted Aβ40 and Aβ42 solutions were centrifuged at 10 000 rpm for 1 min and diluted to the desired concentration with sterilized water. Matrigel (diluted in ReN cell differentiation medium at the volume ratio 1:1) was injected into the 3D AD culture MC, collagen scaffold was then introduced to the collagen scaffold MC, and after Matrigel coating hCMEC/D3 was seeded and the monolayer formed. After monolayer formation, the Aβ40 and Aβ42 solutions were introduced into the ReN cell media MC with ReN cell differentiation medium, after which the ReN cell and BBB chambers were connected.

Fluorescent-Tagged Aβ40 and Aβ42 Treatment for CAA Model: Synthetic human HiLyte Fluor 555-labeled Aβ40 and FAM-labeled Aβ42 (Anaspec, San Jose, CA, USA) were reconstituted in 1% NH4OH and diluted to

the desired concentration of 500 ng mL−1 with ReN cell differentiation media. The reconstituted synthetic human HiLyte Fluor 555-labeled Aβ40 and FAM-labeled Aβ42 were added into the ReN cell media MC and refreshed daily for 6 d.

Immunofluorescent Staining and Image Acquisition: Cells cultured

in the microfluidic system were fixed by incubating with 4% (w/v) paraformaldehyde in PBS for MAP2; GFAP; VE-cad; ZO-1; RAGE; pgp; LRP1; Aβ40/42 antibodies or methanol for claudin-5 antibody for 15 min, and permeabilized with 1% Trion-x100 (Sigma-Aldrich) in PBS for 5 min. After blocking overnight at 4 °C, primary antibodies including anti-MAP2 (1:50, Cell Signaling), anti-GFAP (1:100, Abcam); anti-VE-cadherin (1:100, Enzo Lifesciences); anti-ZO-1 (10 µg mL−1, Invitrogen); anti-claudin-5

(5 µg mL−1, Abcam); anti-RAGE (0.6 mg mL−1; Abcam); anti-pgp, (1:500,

PBS, Sigma-Aldrich); anti-LRP1(1:100, Abcam); and anti-Aβ40/42 (1:100, Cell Signaling) were added and incubated overnight at 4 °C. After washing the MCs with PBS at least two times, secondary antibodies including Alex Fluor-488-conjugated goat-rabbit and goat-mouse IgG (1:200, Molecular Probes); Alex-647-conjugated goat-rabbit and goat-mouse IgG were introduced to the channels and incubated at RT for 2 h, followed by 6-diamidino-2-phenylindole (DAPI, Sigma-Aldrich) for cell nuclei staining.

Intracellular ROS Measurement: Intracellular ROX production was

measured by CellROX Orange Reagent (Life Technologies, City, MD, USA) according to manufacturer’s protocol. At coculture day 3, HCMEC/ D3 cells in the bEC barrier MC were incubated with 5 × 10−6m of CellROX

Orange Reagent (Invitrogen, Carlsbad, CA, USA) for 30 min at 37 °C in the dark, followed by triple washing with prewarmed PBS. Then, cells were examined with a confocal laser scanning microscope (FMV-1000, Olympus, Japan) at an excitation/emission wave-length of 545/565 nm.

Cell Apoptosis Assay: On coculture day 3, HCMEC/D3 cells in the bEC

barrier MC were incubated with 5 × 10−6m of CellEvent Caspase-3/7

Green Detection Reagent (Invitrogen, Carlsbad, CA, USA) with Hoechst for 30 min at 37 °C in the dark and washed twice with prewarmed PBS. Cells were imaged with a confocal laser scanning microscope (FMV-1000, Olympus, Japan) at excitation/emission wave-length of 502/530 nm for CellEvent Caspase-3/7 Green Detection Reagent and 361⁄497 nm for Hoechst.

Real-Time Quantitative Reverse Transcription PCR: Total RNA was isolated

and purified using the RNeasy Mini kit (Qiagen, Chatsworth, CA, USA) according to manufacturer’s instructions. The concentration of total RNA was measured using a Nanodrop 1000 Spectrophotometer. cDNA was synthesized using High-Capacity RNA-to-cDNA Kit (Applied Biosystems, Foster City, CA, USA) according to manufacturer’s protocol. Quantitative Real-time RT-PCR was performed on a 7900HT Fast Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) using TaqMan Fast Advanced Master Mix (Applied Biosystems, Foster City, CA, USA). GAPDH was used as a housekeeping gene. Expressions of the following genes in hCMEC/ D3 cells were quantified using TaqMan Gene Expression Assays (Applied Biosystems, Foster City, CA, USA): GAPDH (Hs03929097_g1), CLDN1 (Hs00221623_m1), CLDN5(Hs00533949_s1), OCLN(Hs00170162_m1), ZO-1(Hs01551861_m1), CDH5 (Hs00901463_m1).

Computational Simulation of Concentration Profile of Aβ in the AD-BBB

Model: Concentration profiles of Aβ within the microfluidic system were simulated using COMSOL Multiphysics 5.2. The changes of the concentration profiles over time were estimated by Fick’s second law

∂ ∂ = ∂∂ 2 2 C t D Cx (2)

where C is concentration and D is the diffusion coefficient. Aβ size was taken as 4 kDa, and the diffusion coefficient of Aβ in media, Matrigel, 2 mg mL−1 collagen, and 5 mg mL−1 collagen were taken from the

literature to be 1.8 × 10−10, 1.24 × 10−10, 0.7 × 10−10, and 0.62 × 10−10 m2 s−1,

respectively.[51–53] The initial value of the 3D ReN culture MC was set to

1.65 × 10−7 mol m−3 considering Aβ production before connecting two

chambers. The rate of Aβ production by AD cells was assumed to be 5 × 10−12m per hour and diffusion through cells was assumed to be

2 × 10−15 m2 s−1 based on the detected Aβ amount from media in ReN

cell media and bEC barrier MC.

BACE1 Inhibitor Treatment: After seeding and differentiating ReN-AD

cells for 2 d, 1 × 10−6m of BACE1 inhibitor (LY2886721, Selleckchem,

Houston, TX) was added with differentiation medium in the ReN media MC. Media was refreshed every day with 1 × 10−6m of BACE1 inhibitor.

ELISA Analysis: The measurement of Aβ40 and Aβ42 was performed using the Meso Scale Discovery (MSD, Rockville, MD) 96-well plate V-PLEX Aβ peptide kits, as outlined in the manufacturer’s protocol. The levels of IFNγ, IL-1β, IL-6, IL-8, and TNFα were measured by MSD 96-well plate Human Pro-Inflammatory V-PLEX Human Pro-Inflammatory Assay kits. MMP-2, MMP-3, and MMP-9 were measured by MSD 96-well plate Human MMP ultrasensitive kits.

Supporting Information

Supporting Information is available from the Wiley Online Library or from the author.

Acknowledgements

Y.S. and S.H.C. contributed equally to this work. This work was supported by the Cure Alzheimer’s Fund (R.D.K. and S.H.C.); the JPB Foundation (R.E.T.); and NIH/NIA grant 1RF1AG048080-01 (D.Y.K. and R.E.T.). Note: “increasing” was corrected to “decreasing” in the sentence “Thrombin was able to pass the leaky BBB and exacerbated neuronal loss in AD, which was prevented by decreasing BBB permeability using pharmacological interventions.” on October 16, 2019 after initial online publication.

(11)

Conflict of Interest

The authors declare no conflict of interest.

Keywords

3D Alzheimer’s disease model, Alzheimer’s disease, blood–brain barrier Received: April 25, 2019 Revised: June 20, 2019 Published online: August 12, 2019

[1] R. E. Tanzi, L. Bertram, Cell 2005, 120, 545.

[2] P. Ballabh, A. Braun, M. Nedergaard, Neurobiol. Dis. 2004, 16, 1. [3] R. Deane, B. V Zlokovic, Curr. Alzheimer Res. 2007, 4, 191.

[4] B. D. Zipser, C. E. Johanson, L. Gonzalez, T. M. Berzin, R. Tavares, C. M. Hulette, M. P. Vitek, V. Hovanesian, E. G. Stopa, Neurobiol.

Aging 2007, 28, 977.

[5] B. Zlokovic, Alzheimer’s Dementia 2015, 11, P114.

[6] D. A. Nation, M. D. Sweeney, A. Montagne, A. P. Sagare, L. M. D’Orazio, M. Pachicano, F. Sepehrband, A. R. Nelson, D. P. Buennagel, M. G. Harrington, T. L. S. Benzinger, A. M. Fagan, J. M. Ringman, L. S. Schneider, J. C. Morris, H. C. Chui, M. Law, A. W. Toga, B. V. Zlokovic, Nat. Med. 2019, 25, 270.

[7] M. D. Sweeney, Z. Zhao, A. Montagne, A. R. Nelson, B. V Zlokovic,

Physiol. Rev. 2019, 99, 21.

[8] M. D. Sweeney, K. Kisler, A. Montagne, A. W. Toga, B. V Zlokovic,

Nat. Neurosci. 2018, 21, 1318.

[9] A. Montagne, Z. Zhao, B. V Zlokovic, J. Exp. Med. 2017, 214, 3151. [10] Y. H. Kim, S. H. Choi, C. D’avanzo, M. Hebisch, C. Sliwinski,

E. Bylykbashi, K. J. Washicosky, J. B. Klee, O. Brüstle, R. E. Tanzi, D. Y. Kim, Nat. Protoc. 2015, 10, 985.

[11] S. H. Choi, Y. H. Kim, M. Hebisch, C. Sliwinski, S. Lee, C. D’Avanzo, H. Chen, B. Hooli, C. Asselin, J. Muffat, J. B. Klee, C. Zhang, B. J. Wainger, M. Peitz, D. M. Kovacs, C. J. Woolf, S. L. Wagner, R. E. Tanzi, D. Y. Kim, Nature 2014, 515, 274.

[12] B. B. Weksler, E. A. Subileau, N. Perrière, P. Charneau, K. Holloway, M. Leveque, H. Tricoire-Leignel, A. Nicotra, S. Bourdoulous, P. Turowski, D. K. Male, F. Roux, J. Greenwood, I. A. Romero, P. O. Couraud, FASEB J. 2005, 19, 1872.

[13] N. A. Rahman, A. N. H. M. Rasil, U. Meyding-Lamade, E. M. Craemer, S. Diah, A. A. Tuah, S. H. Muharram, Brain Res. 2016, 1642, 532. [14] D. J. Selkoe, J. Alzheimer’s Dis. 2001, 3, 75.

[15] C. A Massaad, Curr. Neuropharmacol. 2011, 9, 662.

[16] A. Carrano, J. J. M. Hoozemans, S. M. van der Vies, A. J. M. Rozemuller, J. van Horssen, H. E. de Vries, Antioxid. Redox

Signaling 2011, 15, 1167.

[17] X. Zhu, M. A. Smith, K. Honda, G. Aliev, P. I. Moreira, A. Nunomura, G. Casadesus, P. L. R. Harris, S. L. Siedlak, G. Perry,

J. Neurol. Sci. 2007, 257, 240.

[18] T. Thomas, G. Thomas, C. McLendon, T. Sutton, M. Mullan, Nature

1996, 380, 168.

[19] C. Iadecola, F. Zhang, K. Niwa, C. Eckman, S. K. Turner, E. Fischer, S. Younkin, D. R. Borchelt, K. K. Hsiao, G. A. Carlson, Nat. Neurosci.

1999, 2, 157.

[20] H. Lum, K. A. Roebuck, Am. J. Physiol.: Cell Physiol. 2001, 280, C719. [21] D. S. Bredt, S. H. Snyder, Annu. Rev. Biochem. 1994, 63, 175. [22] J. R. Sowers, N. Engl. J. Med. 2002, 346, 1999.

[23] S. Feng, J. Cen, Y. Huang, H. Shen, L. Yao, Y. Wang, Z. Chen, PLoS

One 2011, 6, e20599.

[24] G. A. Rosenberg, E. Y. Estrada, J. E. Dencoff, Stroke 1998, 29, 2189. [25] A. Tasaki, F. Shimizu, Y. Sano, M. Fujisawa, T. Takahashi, H. Haruki,

M. Abe, M. Koga, T. Kanda, J. Neurol., Neurosurg. Psychiatry 2014,

85, 419.

[26] M. Hernandez-Guillamon, S. Mawhirt, S. Fossati, S. Blais, M. Pares, A. Penalba, M. Boada, P.-O. Couraud, T. A. Neubert, J. Montaner, J. Ghiso, A. Rostagno, J. Biol. Chem. 2010, 285, 27144.

[27] F. H. Duits, M. Hernandez-Guillamon, J. Montaner, J. D. C. Goos, A. Montañola, M. P. Wattjes, F. Barkhof, P. Scheltens, C. E. Teunissen, W. M. van der Flier, J. Alzheimer’s Dis. 2015, 48, 711. [28] N. K.-H. Lim, V. L. Villemagne, C. P. W. Soon, K. M. Laughton,

C. C. Rowe, C. A. McLean, C. L. Masters, G. Evin, Q.-X. Li, J.

Alzhei-mer’s Dis. 2011, 26, 779.

[29] K. D. Rochfort, P. M. Cummins, Biochem. Soc. Trans. 2015, 43, 702. [30] J. Geng, L. Wang, L.-Y. Zhang, C. Qin, Y. Song, Y. Ma, Y. Chen,

S. Chen, Y. Wang, Z. Zhang, G.-Y. Yang, Front. Aging Neurosci. 2018,

10, 129.

[31] J. A. Brown, S. G. Codreanu, M. Shi, S. D. Sherrod, D. A. Markov, M. D. Neely, C. M. Britt, O. S. Hoilett, R. S. Reiserer, P. C. Samson, L. J. McCawley, D. J. Webb, A. B. Bowman, J. A. McLean, J. P. Wikswo, J. Neuroinflammation 2016, 13, 306.

[32] Y.-P. Wei, M. Kita, K. Shinmura, X.-Q. Yan, R. Fukuyama, S. Fushiki, J. Imanishi, J. Interferon Cytokine Res. 2000, 20, 403.

[33] S. Rakshit, B. S. Chandrasekar, B. Saha, E. S. Victor, S. Majumdar, D. Nandi, Biochim. Biophys. Acta 2014, 1843, 2645.

[34] C. T. Ng, L. Y. Fong, Y. Y. Low, J. Ban, M. N. Hakim, Z. Ahmad,

Physiol. Res. 2016, 65, 1053.

[35] C. T. Ng, L. Y. Fong, M. R. Sulaiman, M. A. M. Moklas, Y. K. Yong, M. N. Hakim, Z. Ahmad, J. Interferon Cytokine Res. 2015, 35, 513. [36] I. Chrobak, S. Lenna, L. Stawski, M. Trojanowska, J. Cell. Physiol.

2013, 228, 1774.

[37] P. Grammas, J. M. Martinez, J. Alzheimer’s Dis. 2014, 42, S537. [38] H. Qosa, L. A. Mohamed, S. B. Al Rihani, Y. S. Batarseh,

Q.-V. Duong, J. N. Keller, A. Kaddoumi, J. Alzheimer’s Dis. 2016, 53, 1499. [39] H. Qosa, H. LeVine, J. N. Keller, A. Kaddoumi, Biochim. Biophys.

Acta 2014, 1842, 1806.

[40] M. Li, D.-S. Shang, W.-D. Zhao, L. Tian, B. Li, W.-G. Fang, L. Zhu, S.-M. Man, Y.-H. Chen, J. Immunol. 2009, 182, 5778.

[41] L. M. Tai, K. A. Holloway, D. K. Male, A. J. Loughlin, I. A. Romero, J.

Cell. Mol. Med. 2010, 14, 1101.

[42] M. D. Sweeney, A. P. Sagare, B. V Zlokovic, Nat. Rev. Neurol. 2018,

14, 133.

[43] A. Montagne, S. R. Barnes, M. D. Sweeney, M. R. Halliday, A. P. Sagare, Z. Zhao, A. W. Toga, R. E. Jacobs, C. Y. Liu, L. Amezcua, M. G. Harrington, H. C. Chui, M. Law, B. V. Zlokovic,

Neuron 2015, 85, 296.

[44] R. D. Bell, E. A. Winkler, A. P. Sagare, I. Singh, B. LaRue, R. Deane, B. V Zlokovic, Neuron 2010, 68, 409.

[45] A. Montagne, A. M. Nikolakopoulou, Z. Zhao, A. P. Sagare, G. Si, D. Lazic, S. R. Barnes, M. Daianu, A. Ramanathan, A. Go, E. J. Lawson, Y. Wang, W. J. Mack, P. M. Thompson, J. A. Schneider, J. Varkey, R. Langen, E. Mullins, R. E. Jacobs, B. V. Zlokovic, Nat.

Med. 2018, 24, 326.

[46] J. J. Rodríguez-Arellano, V. Parpura, R. Zorec, A. Verkhratsky,

Neuro-science 2016, 323, 170.

[47] Y. I. Wang, H. E. Abaci, M. L. Shuler, Biotechnol. Bioeng. 2017, 114, 184.

[48] J. L. Mantle, K. H. Lee, Alzheimer’s Dementia 2015, 11, P850. [49] S. G. Canfield, M. J. Stebbins, B. S. Morales, S. W. Asai,

G. D. Vatine, C. N. Svendsen, S. P. Palecek, E. V Shusta, J.

Neuro-chem. 2017, 140, 874.

[50] A. Appelt-Menzel, A. Cubukova, K. Günther, F. Edenhofer, J. Piontek, G. Krause, T. Stüber, H. Walles, W. Neuhaus, M. Metzger,

Stem Cell Rep. 2017, 8, 894.

[51] E. Ciocan, R. Ciocan, in Conf. Proc. IEEE Eng. Med. Biol. Soc., IEEE,

2009, pp. 4925–4928.

[52] J. Waters, PLoS One 2010, 5, e15709.

[53] S. Ramanujan, A. Pluen, T. D. McKee, E. B. Brown, Y. Boucher, R. K. Jain, Biophys. J. 2002, 83, 1650.

Figure

Figure 3.  The increased levels of ROS, MMP-2, and IFNγ in the AD model. a) Relative mean fluorescence intensity of level of intercellular reactive  oxygen species (ROS) in the bEC barrier
Figure 5.  Demonstration of influx of neurotoxins through the impaired BBB and its effects on neural cell damage, and a possible application of drug  evaluation for a BBB enhancer

Références

Documents relatifs

 Early examination of the newborn by the paediatrician in the maternity ward before discharge in the same way as for discharge at 48 h has to be considered, as well as early and

El nombre del método ARDI (Actores, Recursos, Dinámicas, Interacciones) cambio por PARDI para un mejor énfasis en la problemática, el problema o la pregunta (el 'P') a partir de

I. Pinnau, Membrane Technology and Research, Inc., Menlo Park, CA, USA, ipin@mtrinc.com M. Guiver, Institute for Chemical Process and Environmental Technology, National Research

Note, that the three winning families together (all including the deep source) have 95% exceedance probability. c) Exceedance probabilities for each model separately.

Watermarking schemes then are required to respect the Kerckhoffs’ principle [17] (i.e. a secret key is shared between the encoder and the decoder and is the only secret parameter

We now demonstrate the phase-locked temporal modulation of this induced electron current to achieve tunability of resulting CEP-locked THz emission... Temporal characterisation of

We are interested here in the fluctuations of the work injected into the LC when it is driven away from an equilibrium state to another using a small change of the voltage (i.e.

Archaeological and linguistic evidence further suggest that movement and settlement of different communities, particularly, the Bantu, Cushitic and Nilotic groups within the